Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Melinda S. Yates is active.

Publication


Featured researches published by Melinda S. Yates.


Journal of Clinical Oncology | 2011

Phase II Study of Everolimus and Letrozole in Patients With Recurrent Endometrial Carcinoma

Brian M. Slomovitz; Yunyun Jiang; Melinda S. Yates; Pamela T. Soliman; Taren Johnston; Maureen Nowakowski; Charles Levenback; Qian Zhang; Kari L. Ring; Mark F. Munsell; David M. Gershenson; Karen H. Lu; Robert E. Coleman

PURPOSE The phosphoinositol-3 kinase (PI3K) pathway is frequently dysregulated in endometrial cancer (EC). Hormonal manipulation leads to response in some patients with EC, but resistance derived from PI3K pathway activation has been documented. Targeting mammalian target of rapamycin (mTOR) may overcome endocrine resistance. We conducted a two-institution phase II trial of everolimus and letrozole in women with recurrent EC. PATIENTS AND METHODS Patients were considered incurable, had measurable disease, and were treated with up to two prior cytotoxic regimens. Everolimus was administered orally at 10 mg daily and letrozole was administered orally at 2.5 mg daily. Each cycle consisted of 4 weeks of therapy. Patients were treated until progression, toxicity, or complete response (CR). The primary end point was the clinical benefit rate (CBR), which was defined as CR, partial response, or stable disease (≥ 16 weeks) by RECIST 1.0 criteria. Translational studies were performed to correlate biomarkers with response. RESULTS Thirty-eight patients were enrolled (median age, 62 years; range, 24 to 82 years). Thirty-five patients were evaluable for response. The CBR was 40% (14 of 35 patients); the median number of cycles among responders was 15 (range, seven to 29 cycles). The confirmed objective response rate (RR) was 32% (11 of 35 patients; nine CRs and two partial responses; median, 15 cycles; range, eight to 29 cycles). Twenty percent of patients (seven of 35 patients) were taken off treatment after a prolonged CR and at the discretion of the treating clinician. None of the patients discontinued treatment as a result of toxicity. Serous histology was the best predictor of lack of response. Patients with endometrioid histology and CTNNB1 mutations responded well to everolimus and letrozole. CONCLUSION Everolimus plus letrozole results in a high CBR and RR in patients with recurrent EC. Further development of this combination in recurrent endometrioid EC is under way.


Cancer Prevention Research | 2013

Prospective Multicenter Randomized Intermediate Biomarker Study of Oral Contraceptive versus Depo-Provera for Prevention of Endometrial Cancer in Women with Lynch Syndrome

Karen H. Lu; David S. Loose; Melinda S. Yates; Graciela M. Nogueras-Gonzalez; Mark F. Munsell; Lee-may Chen; Henry T. Lynch; Terri L. Cornelison; Stephanie Boyd-Rogers; Mary Rubin; Molly S. Daniels; Peggy Conrad; Andrea Milbourne; David M. Gershenson; Russell Broaddus

Women with Lynch syndrome have a 40% to 60% lifetime risk for developing endometrial cancer, a cancer associated with estrogen imbalance. The molecular basis for endometrial-specific tumorigenesis is unclear. Progestins inhibit estrogen-driven proliferation, and epidemiologic studies have shown that progestin-containing oral contraceptives (OCP) reduce the risk of endometrial cancer by 50% in women at general population risk. It is unknown whether they are effective in women with Lynch syndrome. Asymptomatic women ages 25 to 50 with Lynch syndrome were randomized to receive the progestin compounds Depo-Provera (depo-MPA) or OCP for three months. An endometrial biopsy and transvaginal ultrasound were conducted before and after treatment. Endometrial proliferation was evaluated as the primary endpoint. Histology and a panel of surrogate endpoint biomarkers were evaluated for each endometrial biopsy as secondary endpoints. A total of 51 women were enrolled, and 46 completed treatment. Two of the 51 women had complex hyperplasia with atypia at the baseline endometrial biopsy and were excluded from the study. Overall, both depo-MPA and OCP induced a dramatic decrease in endometrial epithelial proliferation and microscopic changes in the endometrium characteristic of progestin action. Transvaginal ultrasound measurement of endometrial stripe was not a useful measure of endometrial response or baseline hyperplasia. These results show that women with Lynch syndrome do show an endometrial response to short-term exogenous progestins, suggesting that OCP and depo-MPA may be reasonable chemopreventive agents in this high-risk patient population. Cancer Prev Res; 6(8); 774–81. ©2013 AACR.


Cancer Prevention Research | 2011

Microscopic and early-stage ovarian cancers in brca1/2 mutation carriers: Building a model for early BRCA-associated tumorigenesis

Melinda S. Yates; Larissa A. Meyer; Michael T. Deavers; Molly S. Daniels; Elizabeth Keeler; Samuel C. Mok; David M. Gershenson; Karen H. Lu

Risk-reducing salpingo-oophorectomy (RRSO) is the cornerstone of ovarian cancer prevention in BRCA1/2 mutation carriers. Occult fallopian tube and ovarian cancers have been reported in a small percentage of BRCA1/2 mutation carriers undergoing RRSO. Here, we review our single-institution experience with RRSO in BRCA1/2 mutation carriers to characterize cases of microscopic cancers in these patients. At the time of RRSO, 7.9% of BRCA1 mutation carriers were diagnosed with microscopic fallopian tube or ovarian cancers and no cases were diagnosed in BRCA2 mutation carriers. The majority of the microscopic cancers include cases that were confined to the fallopian tubes, although there were also cases involving ovaries only or peritoneal washings only. This suggests that the site of origin may be in the ovary, fallopian tube, or peritoneum for BRCA-associated serous cancers. However, an analysis of early-stage (stages I and II) ovarian and fallopian tube cancers diagnosed in BRCA1/2 mutation carriers confirms that the ovary is a preferred site for tumor growth with 11 of 14 early-stage cancers having a dominant ovarian mass. Overall, these data suggest that cancer initiation may occur in the ovary, fallopian tube, or peritoneum, but tumor growth and progression are favored in the ovary. We present an updated model for BRCA1/2 mutation–associated ovarian and fallopian tube carcinogenesis, which may aid in identifying improved prevention strategies for high-risk women who delay or decline RRSO. Cancer Prev Res; 4(3); 463–70. ©2011 AACR.


Cancer Prevention Research | 2015

Salpingectomy as a Means to Reduce Ovarian Cancer Risk

Mary B. Daly; Charles W. Dresher; Melinda S. Yates; Joanne M. Jeter; Beth Y. Karlan; David S. Alberts; Karen H. Lu

Bilateral salpingo-oophorectomy (BSO) has become the standard-of-care for risk reduction in women at hereditary risk of ovarian cancer. Although this procedure significantly decreases both the incidence of and mortality from ovarian cancer, it affects quality of life, and the premature cessation of ovarian function may have long-term health hazards. Recent advances in our understanding of the molecular pathways of ovarian cancer point to the fallopian tube epithelium as the origin of most high-grade serous cancers (HGSC). This evolving appreciation of the role of the fallopian tube in HGSC has led to the consideration of salpingectomy alone as an option for risk management, especially in premenopausal women. In addition, it is postulated that bilateral salpingectomy with ovarian retention (BSOR), may have a public health benefit for women undergoing benign gynecologic surgery. In this review, we provide the rationale for salpingectomy as an ovarian cancer risk reduction strategy. Cancer Prev Res; 8(5); 342–8. ©2015 AACR. See related commentary by Mark H. Greene, p. 339


Cancer | 2013

Molecular pathogenesis of endometrial cancers in patients with Lynch syndrome

Marilyn Huang; Bojana Djordjevic; Melinda S. Yates; Diana L. Urbauer; Charlotte C. Sun; Jennifer K. Burzawa; Molly S. Daniels; Shannon N. Westin; Russell Broaddus; Karen H. Lu

The authors hypothesized that Lynch syndrome (LS)‐associated endometrial cancer (EC) develops from morphologically normal endometrium that accumulates enough molecular changes to progress through a continuum of hyperplasia to carcinoma, similar to sporadic EC. The primary objective of the current study was to determine whether LS‐associated EC involves progression through a preinvasive lesion. The secondary objective was to identify molecular changes that contribute to endometrial carcinogenesis in patients with LS.


Gynecologic Oncology | 2016

Prospective evaluation of the molecular effects of metformin on the endometrium in women with newly diagnosed endometrial cancer: A window of opportunity study

Pamela T. Soliman; Qian Zhang; Russell Broaddus; Shannon N. Westin; David Iglesias; Mark F. Munsell; Rosemarie Schmandt; Melinda S. Yates; Lois M. Ramondetta; Karen H. Lu

OBJECTIVE Metformin reduces cancer incidence and improves overall survival in diabetic patients. In preclinical studies, metformin decreases endometrial cancer (EC) cell growth by activation of AMPK/mTOR inhibition. We sought to determine the effects of metformin on serum/tumor biomarkers in women with EC. METHODS In this prospective trial, newly diagnosed EC patients underwent pre-treatment blood draw/endometrial biopsy, were administered oral metformin 850mg daily for ≥7days, and underwent post-treatment blood draw/definitive surgery. Pre- and post- serum analyses were performed. Tumor samples were evaluated for changes in AMPK, PI3K/AKT pathway, proliferation, and apoptosis by immunohistochemistry. RESULTS Twenty patients completed the trial. Median age and BMI were 57years (range: 27-67) and 34.5kg/m2 (range: 21.9-50.0). Median duration of metformin was 9.5days (range: 7-24). A majority of women had endometrioid adenocarcinomas (90%) and were early stage (85%). After metformin, there were significant decreases in serum IGF-1 (p=0.046), omentin (p=0.007), insulin (p=0.012), C-peptide (p=0.018), and leptin (p=0.0035). Compared to baseline, post-treatment tissue showed decreased phospho-AKT in 18/20 patients (90%, p=0.0002), decreased phospho-S6rp in 14/20 patients (70%, p=0.057), and decreased phospho-p44/42MAPK in 15/18 patients (83.3%, p=0.0038). There was no difference in Ki67, phospho-ACC, or caspase 3. Changes did not correlate with BMI, grade, or KRAS mutation. CONCLUSION In this prospective window of opportunity study, we demonstrated that relevant serum and molecular changes occur in patients with newly diagnosed EC after a short course of metformin. Ongoing clinical trials will help determine the appropriate role for metformin in the treatment of women with EC.


Gynecologic Oncology | 2014

CGRRF1 as a novel biomarker of tissue response to metformin in the context of obesity

Qian Zhang; Rosemarie Schmandt; Joseph Celestino; Adrienne S. McCampbell; Melinda S. Yates; Diana L. Urbauer; Russell Broaddus; David S. Loose; Gregory L. Shipley; Karen H. Lu

OBJECTIVE Obesity-associated hyperestrogenism and hyperinsulinemia contribute significantly to the pathogenesis of endometrial cancer. We recently demonstrated that metformin, a drug long used for treatment of type 2 diabetes, attenuates both insulin- and estrogen-mediated proliferative signaling in the obese rat endometrium. In this study, we sought to identify tissue biomarkers that may prove clinically useful to predict tissue response for both prevention and therapeutic studies. We identified CGRRF1 (cell growth regulator with ring finger domain 1) as a novel metformin-responsive gene and characterized its possible role in endometrial cancer prevention. METHODS CGRRF1 mRNA expression was evaluated by RT-qPCR in the endometrium of obese and lean rats, and also in normal and malignant human endometrium. CGRRF1 levels were genetically manipulated in endometrial cancer cells, and its effects on proliferation and apoptosis were evaluated by MTT and Western blot. RESULTS CGRRF1 is significantly induced by metformin treatment in the obese rat endometrium. In vitro studies demonstrate that overexpression of CGRRF1 inhibits endometrial cancer cell proliferation. Analysis of human endometrial tumors reveals that CGRRF1 expression is significantly lower in hyperplasia, Grade 1, Grade 2, Grade 3, MMMT, and UPSC endometrial tumors compared to normal human endometrium (p<0.05), suggesting that loss of CGRRF1 is associated with the presence of disease. CONCLUSION CGRRF1 represents a novel, reproducible tissue marker of metformin response in the obese endometrium. Furthermore, our preliminary data suggests that up-regulation of CGRRF1 expression may prove clinically useful in the prevention or treatment of endometrial cancer.


Cancer Prevention Research | 2009

The monkey, the hen, and the mouse: Models to advance ovarian cancer chemoprevention

Karen H. Lu; Melinda S. Yates; Samuel C. Mok

This perspective on Romero et al. (beginning on p. 792 in this issue of the journal) discusses the available animal models of ovarian cancer, including the laying hen, non-human primate, and transgenic rodent models, and their relevance to ovarian cancer chemoprevention studies.


Archive | 2011

Detoxication of Chemical Carcinogens and Chemoprevention

Melinda S. Yates; Thomas W. Kensler

Cancer chemoprevention is an approach that uses natural or synthetic agents, dietary supplements, or foods to block, retard, or even reverse the carcinogenic process. Modulation of the expression of enzymes affecting carcinogen detoxi-cation, such as glutathione S-transferases and UDP-glucuronosyl transferases, is an effective means for cancer chemoprevention. This strategy seeks to alter carcinogen metabolism to facilitate elimination, resulting in protection against mutagenesis, carcinogenesis, and other forms of toxicity mediated by the reactive intermediates of carcinogens. This chapter describes the enzymes involved in carcinogen metabolism and detoxication, along with a discussion of agents that modulate their expression and their effects in animal models of carcinogenesis or human clinical trials.


Archive | 2018

Management of Endometrial Cancer Precursors in Obese Women

J.A. Dottino; Karen H. Lu; Melinda S. Yates

Endometrial cancer can generally be divided into two clinical subtypes, designated as type I and type II disease, each with different risk factors, histology, genetics, treatment and prognosis. Type I endometrial cancer, or low grade endometrioid adenocarcinoma, accounts for 80–90% of new cases of endometrial cancer. Endometrioid endometrial cancer is strongly associated with unopposed estrogen exposure (such as hormone replacement therapy) and obesity. Type II endometrial cancers, including clear cell carcinoma and uterine papillary serous carcinoma, are relatively rare. While the etiology of these rare tumors is not well understood, type II endometrial cancers are not associated with obesity. As such, this chapter will focus on low grade endometrioid (type I) endometrial cancer and its precursor lesions. Due to the close connection between obesity and endometrioid endometrial cancer, obese women are at increased risk for precancerous lesions and low grade endometrial cancers, and consideration is warranted for management of these conditions in the obese population.

Collaboration


Dive into the Melinda S. Yates's collaboration.

Top Co-Authors

Avatar

Karen H. Lu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Qian Zhang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Rosemarie Schmandt

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Joseph Celestino

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Samuel C. Mok

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Russell Broaddus

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Diana L. Urbauer

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

J.B. Pakish

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Molly S. Daniels

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amir A. Jazaeri

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge